<<

Panthi et al. Translational (2018) 7:3 https://doi.org/10.1186/s40035-018-0108-x

REVIEW Open Access , nitric , and neurodegenerative disorders Sandesh Panthi1*, Sumeet Manandhar2 and Kripa Gautam3

Abstract

Hydrogen Sulfide (H2S) and (NO) have become recognized as important gaseous signaling with enormous pharmacological effects, therapeutic value, and central physiological roles. NO is one of the most important regulators of the pathophysiological condition in central (CNS). It is critical in the various functioning of the brain; however, beyond certain concentration/level, it is toxic. H2S was regarded as toxic with the smell like rotten egg. But, it is now regarded as emerging neuroprotectant and neuromodulator. Recently, the use of donors and inhibitors of these signaling molecules have helped us to identify their accurate and precise biological effects. The most abundant of CNS (glutamate) is the initiator of the reaction that forms NO, and H2Sishighly expressed in brain. These molecules are shedding light on the pathogenesis of various neurological disorders. This review is mainly focused on the importance of H2S and NO for normal functioning of CNS. Keywords: Hydrogen sulfide, Nitric oxide, , Gasotransmitters, Gaseous signaling molecules, Neurodegeneration, Neurodegenerative disorders

Background Nobel Prizes for Medicine/ in 1998 [7]. Because The discovery of gaseous signaling molecules like H2S, NO, of its toxic nature and noxious effects, beneficial roles of and (CO) added a new era in biomedical this were previously neglected [8]. CO was the science as these molecules have great importance in mam- second to be discovered as , and it has malian physiology [1]. They have been termed as proved its importance in cardiovascular and neuronal func- ‘gasotransmitters’ as they are either internally produced or tioning [9, 10]. However, the recognition of endogenous synthesized (endogenously) in the organism or are received level of H2S in mammalian tissue, confirmed the existence from the atmosphere and transmit chemical signals thereby of this gasotransmitters [11–13]. Synthesis, functions, and promote or induce various physiological changes inside the role of these gasotransmitters in various physiological mammalian body [2]. The term ‘gasotransmitter’ for these aspect is discussed in previous reviews [14–19]. The pri- molecules was firstly introduced in 2002, and these mary purpose of this review is to highlight the contextual molecules share some common characteristics. They are link between CNS and these gaseous signaling molecules. endogenously produced, enzymatically generated, and their production can be regulated. Gasotransmitters are perme- able to the cell membrane, but their functions inside the NO and CNS body are dependent on their concentration [3, 4]. For better NO is synthesized in CNS from an called as understating and to boost biomedical research in the field L-arginine via an called NO-synthase (NOS) in of gasotransmitters, a society named European Network on equimolar amounts with L-citrulline [20, 21]. There are Gasotransmitters was established in 2011. three different isoforms of NOS which are genetically Nitricoxidewasthefirstgaseousmoleculetobelinked different [22, 23]. Expression of NOS in various part of with its beneficial roles [5]. NO was the molecule of the the brain is shown in Table 1. Because of its ability to year in 1992 [6]injournal‘Science’ and was recognized by passively permeate cell membrane via diffusion, there is no need of receptor binding unlike conventional neuro- transmitter signaling pathways [24]. Its key potential to * Correspondence: [email protected]; [email protected] 1Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand diffuse rapidly in aqueous and environment made it Full list of author information is available at the end of the article unique from other CNS signaling molecules [25].

© The Author(s). 2018 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 2 of 8

Table 1 Expression of NOS in various part of brain glutamatergic neurotransmission and acts as a neuromodula- Isoforms of NOS Expression of NOS tor of excitatory neurotransmitter [28, 31]. Recent researches eNOS Vascular , Choroid plexus also demonstrated the effect of NO on inhibitory GABA- nNOS Neuronal cell bodies especially in thalamus, ergic synaptic transmission [32] via cGMP dependent olfactory bulb, claustrum, amygdala, cortex, suppression of potassium/chloride co-transporter [33]. , hypothalamus Reduction of NO may to the inability of patients to iNOS Glial cells, Macrophages, Neutrophils learn and memorize due to the impairment of long-term potentiation (LTP), as NO is responsible for the increment NO is mainly produced by Neuronal NO-synthase (nNOS) of the synaptic efficiency of pre-synaptic glutamatergic and Endothelial NO-synthase (eNOS) under normal physio- neurons and this increment induces LTP [34]. This gas- logical conditions, but Inducible NO-synthase (iNOS) is only eous signaling molecule also exerts protective role in generated after induction via inflammatory mediators like cy- brain-ischemia as a result of its strong tokines and endotoxins [26]. nNOS was the first isoform to stimulatory effect on angiogenesis and vasodilation [35]. be purified and cloned from the brain [27]. NO diffuses from Hemodynamic and vasodilation activity of NO donors [S- one neuron to another. It is not stored in any kind of synap- nitrosoglutathione (GSNO), S-nitroso-N-acetyl-penicillamine tic vesicles, and its release is independent to membrane (SNAP), sodium nitroprusside (SNP), hexam- depolarization [25, 28]. The generation of NO is similar for methylamine NONOate (MAHMA), propylamine all subtypes of NOS, but the functional regulation and level propylamine NONOate (PAPA), 3-morpholinosydnonimine of production is different. nNOS and eNOS are constitutive (SIN-1), and nitroglycerin (NTG)] has also provided cerebro- forms of NOS, and both rely on the elevation of intracellular vascular neuroprotective role in the various experimental Ca2+ level to initiate NO synthesis. nNOS requires N- model of [36, 37]. NO produced from endothelial cells methyl-D-aspartate (NMDA) receptor activation, and eNOS and adrenergic neurons regulates cerebral blood flow and needs calmodulin-dependent displacement of regulatory pro- smooth muscle tone during conditions like , hyper- teins for NO synthesis. However, iNOS activity is less sensi- capnia, , etc. eNOS mediates the basal release of tive to changes in intracellular Ca2+.But,itcanproducea NO to regulate the cerebral blood flow in various mammals. large amount of NO compared to that of NO associated with This role of NO has been confirmed by various recent eNOS and nNOS [29]. researches using NOS inhibitors [38–40]. The of NO in the target cell is associ- Age-related decrease in cGMP was also linked with in- ated with soluble Guanylate cyclase (GC)/Cyclic-guanosine creased NOS level in one recent study which may be helpful mono phosphate (cGMP) (Fig. 1) or with S-nitrosylation of for anti-aging therapies [41]. NO is also suspected to be a [30]. NO binds with the cGMP producing enzyme crucial molecule to nitrate/S-nitrosylate brain-derived called as GC and expresses its modulating effects as pre-or neurotrophic factor (BDNF) and tropomyosin-related kinase post-synaptic retrograde messenger which facilitates receptor which helps the maintenance of synaptic plasticity

Fig. 1 NO-cGMP signaling pathway: Highly membrane permeable NO binds with NO-soluble Guanylyl cyclase (sGC) which causes conformational changes, and it induces the conversion of GTP-cGMP. cGMP interacts with various intracellular like phosphodiesterase (PDE), cGMP-gated channels (CNG) and protein kinase G (PKG) which stimulates various downstream substrates. Other various pathways are triggered, and many physiological effects starts in cellular level. (Figure adapted and modified from [31]) Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 3 of 8

and LTP [42]. A study has found that NO directly activates reduction of CBF after TBI [56, 57]. Various studies have ryanodine receptor (RyR), which cause the intracellular re- also proved that NO depletion has a critical role in cortical lease of Ca2+ to towards CNS, and it is believed to promote spreading depression, early brain injury, microthrombus the prolonged Ca2+ signaling in the brain. Alike, BDNF and formation, changes in blood flow after subarachnoid tropomyosin-related kinase receptor, this process is also trig- hemorrhage and if we can target this pathway, then it is gered by reversible S-nitrosylation that cause the Ca2+ re- possible to prevent secondary neuronal injury [21]. lease. This whole process is essential for cerebellar synaptic Interestingly, inhaled and intravenous (i.v.) injection of plasticity [43]. Role of NO in maintaining cerebellar synaptic sodium (NO donor) was found to have neuroprotec- plasticity, synaptic transmission efficiency, and cerebellar tive role in mice and rats during cardiac arrest (seen in the LTP are also studied and mentioned [44–46]. clinical model of ischemia/reperfusion) [58, 59]. Not only in NO was also found to have affect in the sleep-wake cycle. the mice model of ischemia, but there are also few studies Intraperitoneal (i.p.) administration of nNOS inhibitor conducted till now which support that inhaled NO or i/v caused the drop in rapid eye movement (REM) and sleep- injected of L-arginine or has protective out- wave-sleep. This relationship between the production of comes in the development of brain after injury, using various NO and sleep is also thought to be linked with various neu- experimental injury models [60–67]. This gasotransmitter is rodegenerative disorders [47]. Researchers linked the role also associated to normalize capillary blood flow, improve- of NO with experimental seizuremodelwhereoverstimula- ment of delivery of [68], prevention and reversal of tion of NMDA receptors is believed to cause the prolonged cerebral vasospasm [21, 66, 67], mitochondrial respiration release of NO [48]. However, the overactivity of same [58], development of healthy brain [21], myelination [69], NMDA receptors leading overproduction of NO may andprotectiveroleafterperipheralnerveinjury[15]. contribute to cell death-initiating various neurodegenerative However, evidence suggests that higher concentration of conditions like Alzheimer’s disease (AD), Parkinson’sdis- NO can impact neuronal death in several ways. Neuronal ease (PD), amyotrophic lateral sclerosis (ALS), stroke and death may occur due to energy depletion-induced inhibition of NOS could be neuroprotective [49]. that may stop mitochondrial respiration or slow inhibition of Brain hypoperfusion and increased vascular oxidative glycolysis. So, the role of NO is particularly dependent on its stress are the common phenomena involved in AD [50, concentration, time-course exposure, and presence/absence 51]. Few researchers also found that abnormal nNOS ex- of ROS at particular level and cells. Thus, being a neuropro- pressions are early symptoms of the AD and cognitive im- tectant at low level, NO might behave as toxicant at higher pairment involved in AD [52, 53]. It was found that concentration [70]. It was found that after the administration imbalance between the nitrotyrosine and all three isoforms of nitro-L- arginine, the infarct size of nNOS-KO mice be- of NOS results in the increased amount of nitrosylation came larger (whereas vascular NO protects after middle and oxidative products in blood and CSF of AD patients cerebral artery occlusion) and NO induced calcium release [54]. Some more physiological roles of the different isoform was found to be involved in neuronal cell death [43, 71]. Be- of NOS are listed in Table 2. causeofthisreasonNOisoftentermedasa“double-edged Stroke (ischemic stroke) is also characterized by inter- sword” [48]. One study revealed the critical role of NO dur- ruption of blood flow in the cerebral artery which ultim- ing neurodegenerative disorders and brain aging can form ately result in ischemia and tissue death. Using the blood-cerebrospinal fluid barrier and this may interfere experimental model of stroke, Huang et al. in 1996 choroid-plexus gateway activity [72]. Similarly, another find- found that eNOS deficient mice has bigger infracts than ing stated NO as the negative player in the progression of wild type [55]. eNOS was also found to maintain the level pathological nature of AD [73]. Recently, another deleterious of cerebral blood flow (CBF) after traumatic brain injury role of NOS with involvement of CNS in mouse model of (TBI). However, eNOS knock-out mice has greater dengue has been discovered [74]. NO is involved in various physiological functioning which is explained earlier in this Table 2 Physiological roles of different isoform of NOS review, however, if the physiological control of signaling Isoforms of NOS Functions pathways involved with NO or NOS fails, then NO and eNOS • Preservation and maintenance of other reactive species (RNS) can cause neuroinflam- ’ brain s microcirculation [21]. mation and neurodegeneration [75]. • Inhibition of platelet aggregation [131].

• Reduction of smooth muscle proliferation [21, 39] H2S and CNS nNOS Important roles in memory formation, CNS blood H2S is a toxic and poisonous gas having an odour of rotten flow, neuronal plasticity, transmission of pain eggs. Alike other gasotransmitters, the physiological role of signals [21]. H2S was overlooked or not paid attention due to its iNOS Response to proinflammatory or [76]. It is the most recent gaseous signaling molecule discov- endotoxins [21]. ered after NO and CO having enormous pathophysiological Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 4 of 8

significance in various disease and conditions [77]. It is neuropsychiatric symptoms which cause neuronal apop- analog of , and because of its weak intermolecular force, tosis, neuronal inflammation (induced by amyloid-β), and it exists in gaseous form [78]. It is synthesized via both increased oxidative stress [90–93]. Level of H2Sinthe enzymatic and non-enzymatic pathways inside mammalian brain of patient with AD is lower than healthy people of tissue, but non-enzymatic route accounts for a small portion. same age [94]. A recent study revealed that in a rat model Cystathionine β-synthase (CBS) and Cystathionine γ-lyase of vascular dementia, plasma H2S level was lower and i.p. (CSE) are two responsible for biosynthesis of H2S injection of NaHS (H2S donor) protected neuronal injury from L-cysteine [79, 80]. 3-mercaptopyruvate sulfurtransfer- and improved behavioral (learning and memory) tests re- ase (3MST) is another enzyme that can generate H2S sults [95]. Another study demonstrated that progression through cys-catabolism pathway. CSE and CBS are localized of AD was abrupted after treatment with spa-water rich of in the cytoplasm of cell, but 3MST is expressed partly in H2Scontent[96]. Role of H2S in the improvement of cog- mitochondria and cytoplasm [81, 82]. A recent study showed nitive functioning, spatial learning and memory [96], and H2S could be produced from D-cysteine via enzyme D- neuroprotective effects [14, 90, 93, 97]isalsoprovidingus amino acid oxidase (DAO) [83, 84]. Non-enzymatically, it hopes against the AD. can be produced from thiosulfate [14]andglucose(via PD is characterized by progressive degeneration of glycolysis) or from phosphogluconate via NADPH oxidase dopaminergic neurons in the of midbrain [85, 86]. Although H2S has beneficial roles in various which is age-related and to the formation of Lewy hematologic diseases, urological disease, cardiovascular func- bodies in soma of residual neurons [77, 98]. Previous stud- tioning and oxidative stress, the effect of H2SinCNShas ies based on animal models found that inhalation or injec- attracted a lot of attention over the past few years [14, 77, tion of H2S donors prevented abnormalities related to PD 87]. Expression of different H2Sproducingenzymesin (microglial activation or motor dysfunction) including various parts of mammalian tissues is listed in Table 3 [88]. neuroprotective, neuromodulatory, and therapeutic roles Important signaling events of H2S in various neuronal cells/ of H2SinPD[99–101]. H2S-mediated anti-oxidative, anti- cell lines are listed below [89]: inflammatory, anti-apoptotic, and pro-survival effects linked with PD is also reviewed in recent paper [102]. 1. Inhibition of monoamine oxidase (via TBIisoneofthemostcommoncausesofdeathamong catecholamines) youth in today’s world and is considered as a public health 2. NMDA potentiation (via glutamate) epidemic. Memory impairment and cognitive dysfunction 3. Cystic fibrosis transmembrane conductance regulator are two immediate effects of TBI whereas rapid and ex- (CTFR) channel activation (via chloride channels) treme production of ROS are also associated with second- 4. KATP and KCa2+ channel activation (via potassium ary neuronal injury after TBI [103–106]. Karimi et al. channels) injected NaHS intraperitoneally and observed neuroprotec- 5. Intracellular calcium mobilisation, L-type and T-type tive effect of H2S in TBI induced impaired memory in rats channel activation (via calcium channels) [103]. Zhang et al. found H2S as a neuromodulator by 6. Supression of various types of neuronal toxicity (via injecting same H2S donor which decreased TBI induced le- oxidative stress) sion volume in brain [107]. NaHS proved to be the neuro- 7. Inhibition of p38-MAPK (via mitogen and protective in various other pathological conditions [108– kinase receptors) 112]. These studies are also supported by recent finding 8. Stimulation of PKA and elevation of cAMP (via PKA) which showed dynamic changes in CBS and H2S levels in various part of the brain in experimental TBI models [107]. AD, a common form of dementia, characterized by Huntington’s Disease (HD) is associated with neurotox- memory impairment, personality changes, and various icity, behavioural changes, impairment of motor coordin- ation, and oxidative stress. Paul et al. showed that there is a reduction of level of CSE in mammalian tissues with HD. Table 3 Expression of different H2S producing enzymes in various parts of mammalian tissues They demonstrated that loss of CSE mediates degeneration of neuronal cells and progression of HD [113, 114]. Studies H2S producing enzymes Expression have shown that patients with Down Syndrome (DS) has CSE Liver, Kidney, Aorta, Ileum but weakly found in brain. the higher level of CBS compared to that of a normal indi- vidual. This overexpression of CBS is believed to be the CBS Liver, Kidney, and Brain (astrocytes) cause of abnormal cognitive ability in children with DS and 3MST Liver, Kidney, Heart, Brain (Purkinje cells of cerebellum, pyramidal neurons of cerebellar may lead to AD in DS adults. Overproduction of H2Sisalso cortex, hippocampus, mitral cells of olfactory associated with ethylmalonic encephalopathy [115]. Some bulb, retinal neurons), Vascular endothelium, other neuroprotective [40, 78, 116–118], neurotransmissive Smooth muscle. role (facilitation of the induction of hippocampal LTP) Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 5 of 8

[119, 120]ofH2Sanditsroleinprotectionofneuronsfrom with each other, their interactions provide beneficial ef- , degeneration [121, 122] and oxidative stress fects on mammalian physiology [14, 124, 128–130]. [121] are also studied extensively and illustrated in Table 4. Conclusions Current understandings and the published data in this field Interrelationship between NO and H S 2 has made clear about their importance in the mammalian Various studies have shown that these gasotransmitters physiology and . There are still many controver- potentiate or antagonize each other’s effect in production, sies surrounding the signaling pathways, beneficial roles, downstream of certain molecular target, and direct and harmful effects of these gasotransmitters. However, chemical interaction [123]. These gasotransmitters share their role in mediation and modulation of cell-to-cell com- the same signaling pathway in the regulation of angiogen- munication is globally accepted fact. Research and studies esis and endothelium dependent vasorelaxation [124]. Few regarding these molecule is still in the preliminary stage in articles demonstrated the common pathway of these gaso- the field of biomedical science, and especially their role in transmitters in the mammalian cardiovascular system [2, CNS is relatively unexplored. Further research also should 17, 125, 126]. Additionally, NO and CO are also found to be focused on their combinatorial effect and signaling path- have link in vasorelaxation and stimulation of calcium sen- ways regarding their antagonistic effect should be disclosed sitive potassium channels [123, 127]. for the development of new therapeutic approaches for Gasotransmitters also have a tendency to compete various neurological disorders. with each other. CO and NO have particular relation- ships with CBS. Research suggests that NO can block Abbreviations the enzymatic activity of H2S via binding with CBS and CBS: Cystathionine β-synthase; CSE: Cystathionine γ-lyase; 3MST: 3-Mercapto CBS has also high preference for CO. If eNOS is im- pyruvate sulfurtransferase; AD: Alzheimer’s disease; ALS: Amyotrophic lateral sclerosis; BDNF: Brain derived neurotrophic factor; cAMP: Cyclic peded, then it cancels out the angiogenic effects of H2S mono phosphate; CBF: Cerebral blood flow; CNS: Central nervous system; whereas blocking H2S significantly lowers the angiogenic CO: Carbon mono-oxide; CTFR: Cystic fibrosis transmembrane conductance regulator; DAO: D-amino acid oxidase; DS: Downs syndrome; effects of NO. CO and H2S act on the same molecular eNOS: Endothelial ; GC: Guanylyl cyclase; H2S: Hydrogen target but have opposite results. Even though H2S, NO, sulfide; HD: Huntington’s disease; i.p.: Intraperitoneal; i.v.: Intravenous; and CO compete and share similar signaling pathways iNOS: Inducible nitric oxide synthase; LTP: Long term potentiation; MAPK: Mitogen-activated protein kinase; NADPH: Nicotinamide adenosine dinucleotide phosphate; NMDA: N-methyl-D-aspartate; nNOS: Neuronal nitric Table 4 Physiological functions of H2S based on its oxide synthase; NO: Nitric oxide; NOS: Nitric oxide synthase; PD: Parkinson’s neuroprotective and neuromodulatory effects disease; PKA: Protein kinase A; REM: Rapid eye movement; RyR: Ryanodine Mode of physiological Evidences receptor; TBI: Traumatic brain injury functions of H2S PD: Inhibits oxygen consumption and Acknowledgements 6-hydroxydopamine evoked NADPH Not Applicable oxidation. Funding Acts on various protein kinases. Not Applicable HD: Upregulation of GSH enzyme and reveals the learning and memory Availability of data and materials problem. Not Applicable AD: Decreases protein oxidation and . Authors’ contributions Reduces -induced toxicity. SP designed and planned about the review topic. SP wrote the manuscript. SM and KG provided intellectual input to the manuscript. All authors read Influences synaptic remodelling. and approved the final manuscript. ALS: Proper regulation of GSH enzyme and reduction of oxidative stress. Ethics approval and consent to participate Not Applicable TBI: Protection via apoptotic and autophagic pathway. Consent for publication Protective effects against neuropathic Not Applicable pain and brain edema.

Neuromodulation • Long term potentiation. Competing interests − The authors declare that they have no competing interests. • CFTR Cl and KATP cycle regulation. • Enhancement of NMDA Author details receptor activity. 1Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand. 2 3 • Regulation of intracellular Ca2+. Department of Pharmacy, Chosun University, Gwangju, South Korea. China Medical University, Shenyang, People’sRepublicofChina. Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 6 of 8

Received: 21 December 2017 Accepted: 31 January 2018 28. Wang H-G, Lu F-M, Jin I, Udo H, Kandel ER, de Vente J, et al. Presynaptic and postsynaptic roles of NO, cGK, and RhoA in long-lasting potentiation and aggregation of synaptic proteins. Neuron. 2005;45:389–403. 29. Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H. Regulation of the References expression of inducible nitric oxide synthase. Nitric Oxide. 2010;23:75–93. 1. Li L, Moore PK. An overview of the biological significance of endogenous 30. Raju K, Ischiropoulos H. Gaseous Signaling in the Central Nervous System. : new roles for old molecules. Portland Press Limited. 2007; Neurosci. 21st Century. 2016;1–16. 2. Fukuto JM, Carrington SJ, Tantillo DJ, Harrison JG, Ignarro LJ, Freeman BA, 31. Chachlaki K, Garthwaite J, Prevot V. The gentle art of saying NO: how nitric et al. Small molecule signaling agents: the integrated and oxide gets things done in the hypothalamus. Nat Rev Endocrinol. 2017;13:521. of nitrogen , oxides of carbon, dioxygen, hydrogen 32. Yamamoto K, Takei H, Koyanagi Y, Koshikawa N, Kobayashi M. Presynaptic sulfide, and their derived species. Chem Res Toxicol. 2012;25:769–93. cell type-dependent regulation of GABAergic synaptic transmission by nitric 3. Wang RUI. Two’s company, three’sa crowd: can H2S be the third oxide in rat insular cortex. Neuroscience. 2015;284:65–77. endogenous gaseous transmitter? FASEB J. 2002;16:1792–8. 33. Yassin L, Radtke-Schuller S, Asraf H, Grothe B, Hershfinkel M, Forsythe ID, 4. Wang R. Signal transduction and the Gasotransmitters: NO, CO, and H2Sin et al. Nitric oxide signaling modulates synaptic inhibition in the superior biology and medicine. Springer Science & Business Media; 2004. paraolivary nucleus (SPN) via cGMP-dependent suppression of KCC2. Front 5. Rajfer J, Aronson WJ, Bush PA, Dorey FJ, Ignarro LJ. Nitric oxide as a Neural Circuits. 2014;8 mediator of relaxation of the corpus cavernosum in response to 34. Puzzo D, Vitolo O, Trinchese F, Jacob JP, Palmeri A, Arancio O. Amyloid-β nonadrenergic, noncholinergic neurotransmission. N Engl J Med Mass peptide inhibits activation of the nitric oxide/cGMP/cAMP-responsive Medical Soc. 1992;326:90–4. element-binding protein pathway during hippocampal synaptic plasticity. 6. Cech TR, Bennett D, Jasny B, Kelner KL, Miller LJ, Szuromi PD, et al. The J Neurosci. 2005;25:6887–97. molecule of the year. Science. 1992;258:1861. 35. Su K, Lin S, Wei J, Lee K, Zhao J, Shyue S, et al. The essential role of 7. Zhou L, Zhu D-Y. Neuronal nitric oxide synthase: structure, subcellular transient receptor potential vanilloid 1 in simvastatin-induced activation of localization, regulation, and clinical implications. Nitric Oxide. 2009;20:223–30. endothelial nitric oxide synthase and angiogenesis. Acta Physiol. 2014;212: 8. Tinajero-Trejo M, Jesse HE, Poole RK. Gasotransmitters, poisons, and 191–204. antimicrobials: it’s a gas, gas, gas! F1000Prime Rep 2013;5. 36. Greco R, Amantea D, Blandini F, Nappi G, Bagetta G, Corasaniti MT, et al. 9. Motterlini R, Otterbein LE. The therapeutic potential of carbon monoxide. Neuroprotective effect of nitroglycerin in a rodent model of ischemic Nat Rev Drug Discov. 2010;9:728–43. stroke: evaluation of Bcl-2 expression. Int Rev Neurobiol. 2007;82:423–35. 10. Carbon MBE. Monoxide: an essential signaling molecule. Med. Organomet 37. Khan M, Jatana M, Elango C, Paintlia AS, Singh AK, Singh I. Cerebrovascular Chem. 2010:247–85. protection by various nitric oxide donors in rats after experimental stroke. 11. Kashfi K, Olson KR. Biology and therapeutic potential of hydrogen sulfide and Nitric Oxide. 2006;15:114–24. hydrogen sulfide-releasing chimeras. Biochem Pharmacol. 2013;85:689–703. 38. Iwanishi K, Watabe H, Hayashi T, Miyake Y, Minato K, Iida H. Influence of 12. Kajimura M, Fukuda R, Bateman RM, Yamamoto T, Suematsu M. Interactions residual oxygen-15-labeled carbon monoxide radioactivity on cerebral of multiple gas-transducing systems: hallmarks and uncertainties of CO, NO, blood flow and oxygen extraction fraction in a dual-tracer autoradiographic – and H2S gas biology. Antioxid Signal. 2010;13:157–92. method. Ann Nucl Med. 2009;23:363 71. 13. Furne J, Saeed A, Levitt MD. Whole tissue hydrogen sulfide concentrations 39. Toda N, Ayajiki K, Okamura T. Cerebral blood flow regulation by nitric oxide: are orders of magnitude lower than presently accepted values. Am J Phys. recent advances. Pharmacol Rev. 2009;61:62–97. 2008;295:1479–85. 40. Kielstein JT, Donnerstag F, Gasper S, Menne J, Kielstein A, Martens- 14. Panthi S, Chung H-J, Jung J, Jeong NY. Physiological importance of Lobenhoffer J, et al. ADMA increases arterial stiffness and decreases cerebral hydrogen sulfide: emerging potent neuroprotector and neuromodulator. blood flow in . Stroke. 2006;37:2024–9. Oxidative Med Cell Longev. 2016;2016 41. Kawamoto EM, Vasconcelos AR, Degaspari S, Böhmer AE, Scavone C, 15. Panthi S, Gautam K. Roles of nitric oxide and ethyl pyruvate after peripheral Marcourakis T. Age-related changes in nitric oxide activity, cyclic GMP, and nerve injury. Inflamm Regen. 2017;37:20. TBARS levels in platelets and erythrocytes reflect the oxidative status in 16. Shefa U, Yeo SG, Kim M-S, Song IO, Jung J, Jeong NY, et al. Role of central nervous system. Age. 2013;35:331–42. Gasotransmitters in oxidative stresses, Neuroinflammation, and neuronal 42. Biojone C, Cabrera Casarotto P, Regiane Joca S, Castren E. Interplay between repair. Biomed Res Int. 2017;2017 nitric oxide and brain-derived neurotrophic factor in neuronal plasticity. CNS 17. Cirino G, Vellecco V, Bucci M. Nitric oxide and hydrogen sulfide: the Neurol Disord Targets. 2015;14:979–87. gasotransmitter paradigm of the vascular system. Br J Pharmacol. 2017; 43. Kakizawa S, Yamazawa T, Chen Y, Ito A, Murayama T, Oyamada H, et al. 18. Qian Y, Matson JB. Gasotransmitter delivery via self-assembling peptides: treating Nitric oxide-induced calcium release via ryanodine receptors regulates diseases with natural signaling gases. Adv Drug Deliv Rev. 2017;110:137–56. neuronal function. EMBO J. 2012;31:417–28. 19. Szabo C. Gasotransmitters in : from pathophysiology to experimental 44. Lev-Ram V, Jiang T, Wood J, Lawrence DS, Tsien RY. Synergies and therapy. Nat Rev Drug Discov. 2016;15:185–203. coincidence requirements between NO, cGMP, and ca 2+ in the induction 20. Knowles RG, Palacios M, Palmer RM, Moncada S. Formation of nitric oxide from of cerebellar long-term depression. Neuron. 1997;18:1025–38. L-arginine in the central nervous system: a transduction mechanism for 45. Lev-Ram V, Wong ST, Storm DR, Tsien RY. A new form of cerebellar long- stimulation of the soluble guanylate cyclase. Proc Natl Acad Sci. 1989;86:5159–62. term potentiation is postsynaptic and depends on nitric oxide but not 21. Garry PS, Ezra M, Rowland MJ, Westbrook J, Pattinson KTS. The role of the cAMP. Proc Natl Acad Sci. 2002;99:8389–93. nitric oxide pathway in brain injury and its treatment—from bench to 46. Katoh A, Kitazawa H, Itohara S, Nagao S. Inhibition of nitric oxide bedside. Exp Neurol. 2015;263:235–43. synthesis and gene knockout of neuronal nitric oxide synthase impaired 22. Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, Stella AMG. adaptation of mouse optokinetic response eye movements. Learn Mem. Nitric oxide in the central nervous system: neuroprotection versus 2000;7:220–6. . Nat Rev Neurosci. 2007;8:766–75. 47. Gautier-Sauvigné S, Colas D, Parmantier P, Clement P, Gharib A, Sarda N, 23. Boissel J-P, Schwarz PM, Förstermann U. Neuronal-type NO synthase: et al. Nitric oxide and sleep. Sleep Med Rev. 2005;9:101–13. transcript diversity and expressional regulation. Nitric Oxide. 1998;2:337–49. 48. Džoljić E, Grabatinić I, Kostić V. Why is nitric oxide important for our brain? 24. Mustafa AK, Gadalla MM, Snyder SH. Signaling by gasotransmitters. Sci Funct Neurol. 2015;30:159–63. Signal. 2009;2:re2. 49. Benarroch EE. Nitric oxide a pleiotropic signal in the nervous system. 25. Garthwaite J, Boulton CL. Nitric oxide signaling in the central nervous Neurology AAN Enterprises. 2011;77:1568–76. system. Annu Rev Physiol. 1995;57:683–706. 50. Nakamura T, Gu Z, Lipton SA. Contribution of glutamatergic signaling to 26. Tieu K, Ischiropoulos H, Przedborski S. Nitric oxide and reactive oxygen nitrosative stress-induced protein misfolding in normal brain aging and species in Parkinson’s disease. IUBMB . 2003;55:329–35. neurodegenerative diseases. Aging Cell. 2007;6:351–9. 27. Bredt DS, Glatt CE, Hwang PM, Fotuhi M, Dawson TM, Snyder SH. Nitric oxide 51. Edwards TM, Rickard NS. New perspectives on the mechanisms through synthase protein and mRNA are discretely localized in neuronal populations of which nitric oxide may affect learning and memory processes. Neurosci the mammalian CNS together with NADPH diaphorase. Neuron. 1991;7:615–24. Biobehav Rev. 2007;31:413–25. Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 7 of 8

52. Lüth H-J, Münch G, Arendt T. Aberrant expression of NOS isoforms in 77. Zhang J, Ding Y, Wang Z, Kong Y, Gao R, Chen G. Hydrogen sulfide therapy Alzheimer’s disease is structurally related to nitrotyrosine formation. Brain in brain diseases: from bench to bedside. Med Gas Res. 2017;7:113. Res. 2002;953:135–43. 78. Qu K, Lee SW, Bian JS, Low C-M, Wong PT-H. Hydrogen sulfide: 53. Malinski T. Nitric oxide and nitroxidative stress in Alzheimer’sdisease. neurochemistry and neurobiology. Neurochem Int. 2008;52:155–65. J Alzheimers Dis. 2007;11:207–18. 79. Chen X, Jhee KH, Kruger WD. Production of the neuromodulator H2S by 54. Virarkar M, Alappat L, Bradford PG, Awad AB. L-arginine and nitric oxide in cystathionine beta-synthase via the condensation of cysteine and CNS function and neurodegenerative diseases. Crit Rev Food Sci Nutr. 2013; homocysteine. J Biol Chem. 2004;279:52082–6. 53:1157–67. 80. Stipanuk MH, Beck PW. Characterization of the enzymic capacity for cysteine 55. Huang Z, Huang PL, Ma J, Meng W, Ayata C, Fishman MC, et al. Enlarged desulphhydration in liver and kidney of the rat. Biochem J. 1982;206:267–77. infarcts in endothelial nitric oxide synthase knockout mice are attenuated 81. Li L, Moore PK. Putative biological roles of hydrogen sulfide in health and by nitro-L-arginine. J Cereb Blood Flow Metab. 1996;16:981–7. disease: a breath of not so fresh air? Trends Pharmacol Sci. 2008;29:84–90. 56. Lundblad C, Grände P-O, Bentzer P. Hemodynamic and histological effects 82. Mikami Y, Shibuya N, Kimura Y, Nagahara N, Ogasawara Y, Kimura H. of traumatic brain injury in eNOS-deficient mice. J Neurotrauma. 2009;26: and dihydrolipoic acid are required for 3-mercaptopyruvate 1953–62. sulfurtransferase to produce hydrogen sulfide. Biochem J. 2011;439:479–85. 57. Robertson CS, Gopinath SP, Valadka AB, Van M, Swank PR, Goodman JC. 83. Tang S, Huang D, An N, Chen D, Zhao D. A novel pathway for the Variants of the endothelial nitric oxide gene and cerebral blood flow after production of H 2 S by DAO in rat jejunum. Neurogastroenterol Motil. 2016; severe traumatic brain injury. J Neurotrauma. 2011;28:727–37. 84. Shibuya N, Koike S, Tanaka M, Ishigami-Yuasa M, Kimura Y, Ogasawara Y, et 58. Dezfulian C, Alekseyenko A, Dave KR, Raval AP, Do R, Kim F, et al. Nitrite al. A novel pathway for the production of hydrogen sulfide from D-cysteine therapy is neuroprotective and safe in cardiac arrest survivors. Nitric Oxide. in mammalian cells. Nat Commun. 2013;4:1366. 2012;26:241–50. 85. Polhemus DJ, Lefer DJ. Emergence of hydrogen sulfide as an endogenous 59. Ichinose F. Improving outcomes after cardiac arrest using NO inhalation. gaseous signaling molecule in . Circ Res. 2014;114:730–7. Trends Cardiovasc Med. 2013;23:52–8. 86. Kolluru GK, Shen X, Bir SC, Kevil CG. Hydrogen sulfide chemical biology: 60. Terpolilli NA, Kim S-W, Thal SC, Kuebler WM, Plesnila N. Inhaled nitric oxide pathophysiological roles and detection. Nitric Oxide. 2013;35:5–20. reduces secondary brain damage after traumatic brain injury in mice. 87. Streeter E, Ng HH, Hart JL. Hydrogen sulfide as a vasculoprotective factor. J Cereb Blood Flow Metab. 2013;33:311–8. Med. Gas Res. 2013;3:9. 61. Terpolilli NA, Kim S-W, Thal SC, Kataoka H, Zeisig V, Nitzsche B, et al. Inhalation 88. Hermann A, Sitdikova GF, Weiger TM. Gasotransmitters: Physiology and of nitric oxide prevents ischemic brain damage in experimental stroke by Pathophysiology. 2012. selective dilatation of collateral arterioles. Circ Res. 2011; Circresaha-111 89. Tan BH, Wong PT-H, Bian J-S. Hydrogen sulfide: a novel signaling molecule 62. Zhang F, White JG, Iadecola C. Nitric oxide donors increase blood flow and in the central nervous system. Neurochem Int. 2010;56:3–10. reduce brain damage in focal ischemia: evidence that nitric oxide is 90. Gong Q-H, Wang Q, Pan L-L, Liu X-H, Huang H, Zhu Y-Z. Hydrogen beneficial in the early stages of cerebral ischemia. J Cereb Blood Flow sulfide attenuates lipopolysaccharide-induced cognitive impairment: a Metab. 1994;14:217–26. pro-inflammatory pathway in rats. Pharmacol Biochem Behav. 2010;96:52–8. 63. Jung K-H, Chu K, Ko S-Y, Lee S-T, Sinn D-I, Park D-K, et al. Early intravenous 91. Tang X, Yang C, Chen J, Yin W, Tian S, Hu B, et al. Effect of hydrogen infusion of sodium nitrite protects brain against in vivo ischemia-reperfusion sulphide on β-amyloid-induced damage in PC12 cells. Clin Exp Pharmacol injury. Stroke. 2006;37:2744–50. Physiol. 2008;35:180–6. 64. Morikawa E, Moskowitz MA, Huang Z, Yoshida T, Irikura K, Dalkara T. L-arginine 92. Fan H, Guo Y, Liang X, Yuan Y, Qi X, Wang M, et al. Hydrogen sulfide infusion promotes nitric oxide-dependent vasodilation, increases regional protects against -peptide induced neuronal injury via cerebral blood flow, and reduces infarction volume in the rat. Stroke. 1994;25: attenuating inflammatory responses in a rat model. J Biomed. 2013;27:296. 429–35. 93. Xuan A, Long D, Li J, Ji W, Zhang M, Hong L, et al. Hydrogen sulfide 65. Li Y-S, Shemmer B, Stone E, Nardi MA, Jonas S, Quartermain D. attenuates spatial memory impairment and hippocampal neuroinflammation in Neuroprotection by inhaled nitric oxide in a murine stroke model is beta-amyloid rat model of Alzheimer’s disease. J Neuroinflammation. 2012;9:202. concentration and duration dependent. Brain Res. 2013;1507:134–45. 94. Eto K, Asada T, Arima K, Makifuchi T, Kimura H. Brain hydrogen sulfide is 66. Fathi AR, Pluta RM, Bakhtian KD, Qi M, Lonser RR. Reversal of cerebral severely decreased in Alzheimer’s disease. Biochem Biophys Res Commun. vasospasm via intravenous sodium nitrite after subarachnoid hemorrhage in 2002;293:1485–8. primates. J Neurosurg. 2011;115:1213–20. 95. Zhang L-M, Jiang C-X, Liu D-W. Hydrogen sulfide attenuates neuronal injury 67. Pluta R, Dejam AA, Gladwin M, Oldfiled E. Nitrite infusions prevent delayed induced by vascular dementia via inhibiting apoptosis in rats. Neurochem cerebral vasospasm in a primate model of subarachnoid hemorrhage. Free Res. 2009;34:1984–92. Radic Biol Med. 2004;37:S81. 96. Giuliani D, Ottani A, Zaffe D, Galantucci M, Strinati F, Lodi R, et al. Hydrogen 68. Shiva S. Nitrite: a physiological store of nitric oxide and modulator of sulfide slows down progression of experimental Alzheimer’s disease by mitochondrial function. Redox Biol. 2013;1:40–4. targeting multiple pathophysiological mechanisms. Neurobiol Learn Mem. 69. Olivier P, Loron G, Fontaine RH, Pansiot J, Dalous J, Thi HP, et al. Nitric oxide 2013;104:82–91. plays a key role in myelination in the developing brain. J Neuropathol Exp 97. Schreier SM, Muellner MK, Steinkellner H, Hermann M, Esterbauer H, Exner Neurol. 2010;69:828–37. M, et al. Hydrogen sulfide scavenges the cytotoxic lipid oxidation product 70. Brown GC. Nitric oxide and neuronal death. Nitric Oxide. 2010;23:153–65. 4-HNE. Neurotox Res. 2010;17:249–56. 71. Huang Z, Huang PL, Panahian N, Dalkara T, Fishman MC, Moskowitz MA. Effects of 98. Kida K, Ichinose F. Hydrogen sulfide and neuroinflammation. Chem cerebral ischemia in mice deficient in neuronal nitric oxide synthase. Sci. 1994:1883. Biochem Pharmacol Hydrog Sulfide. 2015:181–9. 72. Baruch K, Kertser A, Porat Z, Schwartz M. Cerebral nitric oxide represses 99. Kida K, Yamada M, Tokuda K, Marutani E, Kakinohana M, Kaneki M, et al. Inhaled choroid plexus NFκB-dependent gateway activity for leukocyte trafficking. hydrogen sulfide prevents neurodegeneration and movement disorder in a EMBO J. 2015;34:1816–28. mouse model of Parkinson’s disease. Antioxid Redox Signal. 2011;15:343–52. 73. P Fernandez A, Pozo-Rodrigalvarez A, Serrano J, Martinez-Murillo R. Nitric 100. Hu L, Lu M, Tiong CX, Dawe GS, Hu G, Bian J. Neuroprotective effects of oxide: target for therapeutic strategies in Alzheimer’s disease. Curr Pharm hydrogen sulfide on Parkinson’s disease rat models. Aging Cell. 2010;9:135–46. Des. 2010;16:2837–50. 101. Xie L, Hu L-F, Teo XQ, Tiong CX, Tazzari V, Sparatore A, et al. Therapeutic 74. de Souza KPR, Silva EG, de Oliveira Rocha ES, Figueiredo LB, de Almeida- effect of hydrogen sulfide-releasing L-Dopa derivative ACS84 on 6-OHDA- Leite CM, Arantes RME, et al. Nitric oxide synthase expression correlates induced Parkinson’s disease rat model. PLoS One. 2013;8:e60200. with death in an experimental mouse model of dengue with CNS 102. Cao X, Cao L, Ding L, Bian J. A new hope for a devastating disease: involvement. Virol J. 2013;10:267. hydrogen sulfide in Parkinson’s disease. Mol Neurobiol. 2017:1–11. 75. Yuste JE, Tarragon E, Campuzano CM, Ros-Bernal F. Implications of glial 103. Karimi SA, Hosseinmardi N, Janahmadi M, Sayyah M, Hajisoltani R. The nitric oxide in neurodegenerative diseases. Front Cell Neurosci. 2015;9 protective effect of hydrogen sulfide (H2S) on traumatic brain injury (TBI) 76. Kimura H, Nagai Y, Umemura K, Kimura Y. Physiological roles of hydrogen induced memory deficits in rats. Brain Res Bull. 2017;134:177–82. sulfide: synaptic modulation, neuroprotection, and smooth muscle 104. Borgens R. Ben, Liu-Snyder P. Understanding secondary injury Q Rev Biol. relaxation. Antioxid Redox Signal. 2005;7:795–803. 2012;87:89–127. Panthi et al. Translational Neurodegeneration (2018) 7:3 Page 8 of 8

105. Mustafa AG, Alshboul OA. Pathophysiology of traumatic brain injury. Neurosci. 2013;18:222–34. 106. Homsi S, Federico F, Croci N, Palmier B, Plotkine M, Marchand-Leroux C, et al. Minocycline effects on : relations with inflammatory and oxidative stress markers following traumatic brain injury in mice. Brain Res. 2009;1291:122–32. 107. Zhang M, Shan H, Wang T, Liu W, Wang Y, Wang L, et al. Dynamic change of hydrogen sulfide after traumatic brain injury and its effect in mice. Neurochem Res. 2013;38:714–25. 108. Dai H-B, Xu M-M, Lv J, Ji X-J, Zhu S-H, Ma R-M, et al. Mild combined with hydrogen sulfide treatment during resuscitation reduces hippocampal neuron apoptosis via NR2A, NR2B, and PI3K-Akt signaling in a rat model of cerebral ischemia-reperfusion injury. Mol Neurobiol. 2016;53:4865–73. 109. Li T, Liu H, Xue H, Zhang J, Han X, Yan S, et al. Neuroprotective effects of hydrogen sulfide against early brain injury and secondary cognitive deficits following subarachnoid hemorrhage. Brain Pathol. 2016; 110. Yonezawa D, Sekiguchi F, Miyamoto M, Taniguchi E, Honjo M, Masuko T, et al. A protective role of hydrogen sulfide against oxidative stress in rat gastric mucosal epithelium. Toxicology. 2007;241:11–8. 111. Chu Q-J, He L, Zhang W, Liu C-L, Ai Y-Q, Zhang Q. Hydrogen sulfide attenuates surgical trauma-induced inflammatory response and cognitive deficits in mice. J Surg Res. 2013;183:330–6. 112. Zhang M, Shan H, Chang P, Wang T, Dong W, Chen X, et al. Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice. PLoS One. 2014;9:e87241. 113. Paul BD, Sbodio JI, Xu R, Vandiver MS, Cha JY, Snowman AM, et al. Cystathionine gamma-lyase deficiency mediates neurodegeneration in Huntington’s disease. Nature. 2014;509:96–100. 114. Paul BD, Snyder SH. Role Of neuronal signaling effector hydrogen sulfide (H2S) and sulfhydration in Huntington’s disease. FASEB J. 2016;30:1271–6. 115. Kimura H, Shibuya N, Kimura Y. Hydrogen sulfide is a signaling molecule and a cytoprotectant. Antioxid Redox Signal. 2012;17:45–57. 116. Deng J, Lei C, Chen Y, Fang Z, Yang Q, Zhang H, et al. Neuroprotective gases–fantasy or reality for clinical use? Prog Neurobiol. 2014;115:210–45. 117. Mikami Y, Shibuya N, Kimura Y, Nagahara N, Yamada M, Kimura H. Hydrogen sulfide protects the retina from light-induced degeneration by the modulation of Ca2+ influx. J Biol Chem. 2011;286:39379–86. 118. Kimura H. The physiological role of hydrogen sulfide and beyond. Nitric Oxide. 2014;41:4–10. 119. Abe K, Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci. 1996;16:1066–71. 120. Lee SW, Hu Y-S, Hu L-F, Lu Q, Dawe GS, Moore PK, et al. Hydrogen sulphide regulates calcium in microglial cells. Glia. 2006;54:116–24. 121. Geng B, Chang L, Pan C, Qi Y, Zhao J, Pang Y, et al. Endogenous hydrogen sulfide regulation of myocardial injury induced by isoproterenol. Biochem Biophys Res Commun. 2004;318:756–63. 122. White BJO. The vascular effects of hydrogen sulphide. 2012. 123. Wang R. Shared signaling pathways among gasotransmitters. Proc Natl Acad Sci. 2012;109:8801–2. 124. Coletta C, Papapetropoulos A, Erdelyi K, Olah G, Módis K, Panopoulos P, et al. Hydrogen sulfide and nitric oxide are mutually dependent in the regulation of angiogenesis and endothelium-dependent vasorelaxation. Proc Natl Acad Sci. 2012;109:9161–6. 125. Olson KRA. Practical look at the chemistry and biology of hydrogen sulfide. Antioxid Redox Signal. 2012;17:32–44. 126. Nagpure BV, Interaction BJ-S. Of hydrogen sulfide with nitric oxide in the cardiovascular system. Oxidative Med Cell Longev. 2015;2016 127. Furchgott RF, Jothianandan D. Endothelium-dependent and-independent vasodilation involving cyclic GMP: relaxation induced by nitric oxide, carbon Submit your next manuscript to BioMed Central monoxide and light. J Vasc Res. 1991;28:52–61. 128. Taoka S, Banerjee R. Characterization of NO binding to cystathionine and we will help you at every step: β-synthase:: Possible implications of the effects of CO and NO binding to • We accept pre-submission inquiries the human enzyme. J Inorg Biochem. 2001;87:245–51. 129. Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of H(2)S as a novel • Our selector tool helps you to find the most relevant journal endogenous gaseous K(ATP) channel opener. EMBO J. 2001;20:6008–16. • We provide round the clock customer support 130. Beltowski J, Jamroz-Wiśnniewska A. Hydrogen sulfide and endothelium- • Convenient online submission dependent vasorelaxation. Molecules. 2014;19:21506–28. 131. Broos K, Feys HB, De Meyer SF, Vanhoorelbeke K, Deckmyn H. Platelets at • Thorough work in primary hemostasis. Blood Rev. 2011;25:155–67. • Inclusion in PubMed and all major indexing services • Maximum visibility for your research

Submit your manuscript at www.biomedcentral.com/submit